Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(94): eadk0092, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38579014

ABSTRACT

The transition from immunoglobulin M (IgM) to affinity-matured IgG antibodies is vital for effective humoral immunity. This is facilitated by germinal centers (GCs) through affinity maturation and preferential maintenance of IgG+ B cells over IgM+ B cells. However, it is not known whether the positive selection of the different Ig isotypes within GCs is dependent on specific transcriptional mechanisms. Here, we explored IgG1+ GC B cell transcription factor dependency using a CRISPR-Cas9 screen and conditional mouse genetics. We found that MIZ1 was specifically required for IgG1+ GC B cell survival during positive selection, whereas IgM+ GC B cells were largely independent. Mechanistically, MIZ1 induced TMBIM4, an ancestral anti-apoptotic protein that regulated inositol trisphosphate receptor (IP3R)-mediated calcium (Ca2+) mobilization downstream of B cell receptor (BCR) signaling in IgG1+ B cells. The MIZ1-TMBIM4 axis prevented mitochondrial dysfunction-induced IgG1+ GC cell death caused by excessive Ca2+ accumulation. This study uncovers a unique Ig isotype-specific dependency on a hitherto unidentified mechanism in GC-positive selection.


Subject(s)
B-Lymphocytes , Immunoglobulin G , Membrane Proteins , Animals , Mice , Germinal Center , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Signal Transduction , Membrane Proteins/metabolism
2.
Nature ; 621(7980): 813-820, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37587341

ABSTRACT

Disruption of the lung endothelial-epithelial cell barrier following respiratory virus infection causes cell and fluid accumulation in the air spaces and compromises vital gas exchange function1. Endothelial dysfunction can exacerbate tissue damage2,3, yet it is unclear whether the lung endothelium promotes host resistance against viral pathogens. Here we show that the environmental sensor aryl hydrocarbon receptor (AHR) is highly active in lung endothelial cells and protects against influenza-induced lung vascular leakage. Loss of AHR in endothelia exacerbates lung damage and promotes the infiltration of red blood cells and leukocytes into alveolar air spaces. Moreover, barrier protection is compromised and host susceptibility to secondary bacterial infections is increased when endothelial AHR is missing. AHR engages tissue-protective transcriptional networks in endothelia, including the vasoactive apelin-APJ peptide system4, to prevent a dysplastic and apoptotic response in airway epithelial cells. Finally, we show that protective AHR signalling in lung endothelial cells is dampened by the infection itself. Maintenance of protective AHR function requires a diet enriched in naturally occurring AHR ligands, which activate disease tolerance pathways in lung endothelia to prevent tissue damage. Our findings demonstrate the importance of endothelial function in lung barrier immunity. We identify a gut-lung axis that affects lung damage following encounters with viral pathogens, linking dietary composition and intake to host fitness and inter-individual variations in disease outcome.


Subject(s)
Endothelial Cells , Lung , Orthomyxoviridae Infections , Receptors, Aryl Hydrocarbon , Animals , Humans , Mice , Apelin/metabolism , Diet , Endothelial Cells/metabolism , Endothelium/cytology , Endothelium/metabolism , Epithelial Cells/metabolism , Erythrocytes/metabolism , Influenza, Human/immunology , Influenza, Human/metabolism , Intestines/metabolism , Leukocytes/metabolism , Ligands , Lung/immunology , Lung/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Receptors, Aryl Hydrocarbon/metabolism
3.
Nat Commun ; 14(1): 721, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36781848

ABSTRACT

Epithelial tissues provide front-line barriers shielding the organism from invading pathogens and harmful substances. In the airway epithelium, the combined action of multiciliated and secretory cells sustains the mucociliary escalator required for clearance of microbes and particles from the airways. Defects in components of mucociliary clearance or barrier integrity are associated with recurring infections and chronic inflammation. The timely and balanced differentiation of basal cells into mature epithelial cell subsets is therefore tightly controlled. While different growth factors regulating progenitor cell proliferation have been described, little is known about the role of metabolism in these regenerative processes. Here we show that basal cell differentiation correlates with a shift in cellular metabolism from glycolysis to fatty acid oxidation (FAO). We demonstrate both in vitro and in vivo that pharmacological and genetic impairment of FAO blocks the development of fully differentiated airway epithelial cells, compromising the repair of airway epithelia. Mechanistically, FAO links to the hexosamine biosynthesis pathway to support protein glycosylation in airway epithelial cells. Our findings unveil the metabolic network underpinning the differentiation of airway epithelia and identify novel targets for intervention to promote lung repair.


Subject(s)
Epithelial Cells , Respiratory System , Epithelium/metabolism , Epithelial Cells/metabolism , Cell Differentiation/physiology , Fatty Acids/metabolism , Respiratory Mucosa/metabolism
4.
Sci Transl Med ; 14(655): eabn3715, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35895836

ABSTRACT

Several variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged during the current coronavirus disease 2019 (COVID-19) pandemic. Although antibody cross-reactivity with the spike glycoproteins (S) of diverse coronaviruses, including endemic common cold coronaviruses (HCoVs), has been documented, it remains unclear whether such antibody responses, typically targeting the conserved S2 subunit, contribute to protection when induced by infection or through vaccination. Using a mouse model, we found that prior HCoV-OC43 S-targeted immunity primes neutralizing antibody responses to otherwise subimmunogenic SARS-CoV-2 S exposure and promotes S2-targeting antibody responses. Moreover, vaccination with SARS-CoV-2 S2 elicited antibodies in mice that neutralized diverse animal and human alphacoronaviruses and betacoronaviruses in vitro and provided a degree of protection against SARS-CoV-2 challenge in vivo. Last, in mice with a history of SARS-CoV-2 Wuhan-based S vaccination, further S2 vaccination induced broader neutralizing antibody response than booster Wuhan S vaccination, suggesting that it may prevent repertoire focusing caused by repeated homologous vaccination. These data establish the protective value of an S2-targeting vaccine and support the notion that S2 vaccination may better prepare the immune system to respond to the changing nature of the S1 subunit in SARS-CoV-2 variants of concern, as well as to future coronavirus zoonoses.


Subject(s)
COVID-19 Vaccines , COVID-19 , Coronavirus OC43, Human , Spike Glycoprotein, Coronavirus , Animals , Antibodies, Neutralizing , Antibodies, Viral , Broadly Neutralizing Antibodies , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Coronavirus OC43, Human/immunology , Humans , Mice , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/immunology , Vaccination
5.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34473195

ABSTRACT

Cytokine-mediated immune-cell recruitment and inflammation contribute to protection in respiratory virus infection. However, uncontrolled inflammation and the "cytokine storm" are hallmarks of immunopathology in severe infection. Cytokine storm is a broad term for a phenomenon with diverse characteristics and drivers, depending on host genetics, age, and other factors. Taking advantage of the differential use of virus-sensing systems by different cell types, we test the hypothesis that specifically blocking TLR7-dependent, immune cell-produced cytokines reduces influenza-related immunopathology. In a mouse model of severe influenza characterized by a type I interferon (IFN-I)-driven cytokine storm, TLR7 antagonist treatment leaves epithelial antiviral responses unaltered but acts through pDCs and monocytes to reduce IFN-I and other cytokines in the lung, thus ameliorating inflammation and severity. Moreover, even in the absence of IFN-I signaling, TLR7 antagonism reduces inflammation and mortality driven by monocyte-produced chemoattractants and neutrophil recruitment into the infected lung. Hence, TLR7 antagonism reduces diverse types of cytokine storm in severe influenza.


Subject(s)
Antiviral Agents/pharmacology , Interferon Type I/pharmacology , Membrane Glycoproteins/metabolism , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/metabolism , Toll-Like Receptor 7/metabolism , Animals , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Host-Pathogen Interactions/drug effects , Humans , Lung/drug effects , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Monocytes/metabolism
6.
Cell Rep ; 28(1): 245-256.e4, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31269444

ABSTRACT

Type I interferon (IFNα/ß) pathways are fine-tuned to elicit antiviral protection while minimizing immunopathology; however, the initiating stimuli, target tissues, and underlying mechanisms are unclear. Using models of physiological and dysregulated IFNα/ß receptor (IFNAR1) surface expression, we show here that IFNAR1-dependent signals set the steady-state IFN signature in both hematopoietic and stromal cells. Increased IFNAR1 levels promote a lung environment refractory to early influenza virus replication by elevating the baseline interferon signature. Commensal microbiota drive the IFN signature specifically in lung stroma, as shown by antibiotic treatment and fecal transplantation. Bone marrow chimera experiments identify lung stromal cells as crucially important for early antiviral immunity and stroma-immune cell interaction for late antiviral resistance. We propose that the microbiota-driven interferon signature in lung epithelia impedes early virus replication and that IFNAR1 surface levels fine-tune this signature. Our findings highlight the interplay between bacterial and viral exposure, with important implications for antibiotic use.


Subject(s)
Anti-Bacterial Agents/pharmacology , Influenza A virus , Influenza, Human/immunology , Influenza, Human/microbiology , Lung/immunology , Microbiota/immunology , Receptor, Interferon alpha-beta/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line , Chimera/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Fecal Microbiota Transplantation , Gene Expression Regulation, Viral/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology , Humans , Influenza A virus/growth & development , Influenza A virus/immunology , Influenza, Human/drug therapy , Influenza, Human/pathology , Interferon Type I/metabolism , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Lung/drug effects , Lung/microbiology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Seq , Receptor, Interferon alpha-beta/genetics , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/microbiology , Stromal Cells/virology
7.
J Invest Dermatol ; 139(1): 157-166, 2019 01.
Article in English | MEDLINE | ID: mdl-30048652

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) propagates within and between individuals via cell-to-cell transmission, and primary infection typically occurs across juxtaposed mucosal surfaces during breastfeeding or sexual intercourse. It is therefore likely that dendritic cells (DCs) are among the first potential targets for HTLV-1. However, it remains unclear how DCs contribute to virus transmission and dissemination in the early stages of infection. We show that an HTLV-1-infected cell line (MT-2) and naturally infected CD4+ T cells transfer p19+ viral particles to the surface of allogeneic DCs via cell-to-cell contacts. Similarly organized cell-to-cell contacts also facilitate DC-mediated transfer of HTLV-1 to autologous CD4+ T cells. These findings shed light on the cellular structures involved in anterograde and retrograde transmission and suggest a key role for DCs in the natural history and pathogenesis of HTLV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Dendritic Cells/virology , Human T-lymphotropic virus 1/physiology , Leukemia, T-Cell/pathology , Virus Replication , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Humans , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/virology , Microscopy, Electron, Scanning , Tumor Cells, Cultured
8.
J Invest Dermatol ; 136(10): 1981-1989, 2016 10.
Article in English | MEDLINE | ID: mdl-27375111

ABSTRACT

Sterile alpha motif (SAM) and histidine-aspartic (HD) domains protein 1 (SAMHD1) was previously identified as a critical post-entry restriction factor to HIV-1 infection in myeloid dendritic cells. Here we show that SAMHD1 is also expressed in epidermis-isolated Langerhans cells (LC), but degradation of SAMHD1 does not rescue HIV-1 or vesicular stomatitis virus G-pseudotyped lentivectors infection in LC. Strikingly, using Langerhans cells model systems (mutz-3-derived LC, monocyte-derived LC [MDLC], and freshly isolated epidermal LC), we characterize previously unreported post-entry restriction activity to HIV-1 in these cells, which acts at HIV-1 reverse transcription, but remains independent of restriction factors SAMHD1 and myxovirus resistance 2 (MX2). We demonstrate that transforming growth factor-ß signaling confers this potent HIV-1 restriction in MDLC during their differentiation and blocking of mothers against decapentaplegic homolog 2 (SMAD2) signaling in MDLC restores cells' infectivity. Interestingly, maturation of MDLC with a toll-like receptor 2 agonist or transforming growth factor-α significantly increases cells' susceptibility to HIV-1 infection, which may explain why HIV-1 acquisition is increased during coinfection with sexually transmitted infections. In conclusion, we report a SAMHD1-independent post-entry restriction in MDLC and LC isolated from epidermis, which inhibits HIV-1 replication. A better understanding of HIV-1 restriction and propagation from LC to CD4(+) T cells may help in the development of new microbicides or vaccines to curb HIV-1 infection at its earliest stages during mucosal transmission.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Langerhans Cells/virology , Monomeric GTP-Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Cell Line , Humans , Monocytes/metabolism , Myxovirus Resistance Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1 , Transforming Growth Factor alpha/metabolism , Virus Replication/physiology
9.
J Invest Dermatol ; 135(11): 2565-2567, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26548487

ABSTRACT

Herpes simplex virus (HSV-1) is a major cause of viral skin infection in humans. Klenner and colleagues now show that the epidermal receptor activator of NFκB ligand (RANKL) is critical for the induction of anti-viral CD8(+) effector T cells (CTL) during cutaneous HSV-1 infection. Activation via RANKL prevents Langerhans cell apoptosis, thus leading to enhanced antigen transport to regional lymph nodes, increasing the CTL-priming capacity of lymph node dendritic cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/immunology , Langerhans Cells/immunology , RANK Ligand/immunology , Receptor Activator of Nuclear Factor-kappa B/immunology , Animals , Humans
10.
J Virol ; 88(21): 12202-12, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25122777

ABSTRACT

UNLABELLED: Vector-borne flaviviruses, such as tick-borne encephalitis virus (TBEV), West Nile virus, and dengue virus, cause millions of infections in humans. TBEV causes a broad range of pathological symptoms, ranging from meningitis to severe encephalitis or even hemorrhagic fever, with high mortality. Despite the availability of an effective vaccine, the incidence of TBEV infections is increasing. Not much is known about the role of the innate immune system in the control of TBEV infections. Here, we show that the type I interferon (IFN) system is essential for protection against TBEV and Langat virus (LGTV) in mice. In the absence of a functional IFN system, mice rapidly develop neurological symptoms and succumb to LGTV and TBEV infections. Type I IFN system deficiency results in severe neuroinflammation in LGTV-infected mice, characterized by breakdown of the blood-brain barrier and infiltration of macrophages into the central nervous system (CNS). Using mice with tissue-specific IFN receptor deletions, we show that coordinated activation of the type I IFN system in peripheral tissues as well as in the CNS is indispensable for viral control and protection against virus induced inflammation and fatal encephalitis. IMPORTANCE: The type I interferon (IFN) system is important to control viral infections; however, the interactions between tick-borne encephalitis virus (TBEV) and the type I IFN system are poorly characterized. TBEV causes severe infections in humans that are characterized by fever and debilitating encephalitis, which can progress to chronic illness or death. No treatment options are available. An improved understanding of antiviral innate immune responses is pivotal for the development of effective therapeutics. We show that type I IFN, an effector molecule of the innate immune system, is responsible for the extended survival of TBEV and Langat virus (LGTV), an attenuated member of the TBE serogroup. IFN production and signaling appeared to be essential in two different phases during infection. The first phase is in the periphery, by reducing systemic LGTV replication and spreading into the central nervous system (CNS). In the second phase, the local IFN response in the CNS prevents virus-induced inflammation and the development of encephalitis.


Subject(s)
Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/mortality , Interferon Type I/immunology , Animals , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interferon/deficiency , Survival Analysis
11.
PLoS Pathog ; 10(3): e1003999, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24675692

ABSTRACT

The innate immune system protects cells against invading viral pathogens by the auto- and paracrine action of type I interferon (IFN). In addition, the interferon regulatory factor (IRF)-1 can induce alternative intrinsic antiviral responses. Although both, type I IFN and IRF-1 mediate their antiviral action by inducing overlapping subsets of IFN stimulated genes, the functional role of this alternative antiviral action of IRF-1 in context of viral infections in vivo remains unknown. Here, we report that IRF-1 is essential to counteract the neuropathology of vesicular stomatitis virus (VSV). IFN- and IRF-1-dependent antiviral responses act sequentially to create a layered antiviral protection program against VSV infections. Upon intranasal infection, VSV is cleared in the presence or absence of IRF-1 in peripheral organs, but IRF-1-/- mice continue to propagate the virus in the brain and succumb. Although rapid IFN induction leads to a decline in VSV titers early on, viral replication is re-enforced in the brains of IRF-1-/- mice. While IFN provides short-term protection, IRF-1 is induced with delayed kinetics and controls viral replication at later stages of infection. IRF-1 has no influence on viral entry but inhibits viral replication in neurons and viral spread through the CNS, which leads to fatal inflammatory responses in the CNS. These data support a temporal, non-redundant antiviral function of type I IFN and IRF-1, the latter playing a crucial role in late time points of VSV infection in the brain.


Subject(s)
Interferon Regulatory Factor-1/immunology , Neurons/virology , Vesicular Stomatitis/immunology , Virus Replication/physiology , Animals , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Immunohistochemistry , Interferon Regulatory Factor-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/pathology , Vesiculovirus/physiology
12.
Exp Gerontol ; 54: 101-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24462805

ABSTRACT

Poor immune protection upon vaccination is a critical determinant of immunosenescence. Latent Cytomegalovirus (CMV) infection has been associated with poor antibody responses to vaccination, but a causative role for CMV in the poor immune response requires experimental evidence and thus could not be confirmed in clinical studies. To test the hypothesis that latent CMV infection causes poor antibody responses, we infected young or adult mice with mouse CMV and challenged them with Vesicular stomatitis virus (VSV) at 15 or 18months of age. Latent, but not primary infection with mouse CMV resulted in diminished neutralizing titers of the serum IgG fraction at day 7 post challenge, which recovered by day 14 post challenge. This phenomenon was specific for mice infected with mouse CMV, but not mice infected with other herpesviruses, like murine herpesvirus-68 or herpes simplex virus type 1, or mice infected with non-persistent viruses, such as influenza or Vaccinia virus. Hence, our data indicate a delay in IgG class-switch that was specific for the CMV infection. Herpesviral infections did not change the B-cell memory compartment, and increased the size of the effector-memory subset of blood CD4 T-cells only when administered in combination. Furthermore, CD4 T-cell response to VSV infection was maintained in latently infected mice. Therefore, our results argue that latent CMV infection impairs B-cell, but not T-cell responses to a challenge with VSV and delays antibody class-switch by a mechanism which may be independent of T-cell help.


Subject(s)
Antibodies, Viral/immunology , Herpesviridae Infections/immunology , Muromegalovirus/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Flow Cytometry , Immunity, Humoral/immunology , Immunoglobulin G/immunology , Immunologic Memory/immunology , Lymph Nodes/virology , Mice, Inbred C57BL , Mice, Inbred CBA
13.
Gastroenterology ; 145(6): 1414-23.e1, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973921

ABSTRACT

BACKGROUND & AIMS: Current treatment strategies for hepatitis C virus (HCV) infection include pegylated interferon (IFN)-alfa and ribavirin. Approximately 50% of patients control HCV infection after treatment, but the broad range of patients' outcomes and responses to treatment, among all genotypes, indicates a role for host factors. Although the IFN system is important in limiting HCV replication, the virus has evolved mechanisms to circumvent the IFN response. However, direct, IFN-independent antiviral processes also might help control HCV replication. We examined the role of IFN-independent responses against HCV replication. METHODS: We analyzed replication of the subgenomic JFH1 replicon in embryonic fibroblasts and primary hepatocytes from mice with disruptions in genes encoding factors in the IFN-dependent and alternative antiviral pathways (signal transducers and activators of transcription 1 [STAT1], protein kinase R, interferon regulatory factors (IRF) IRF-1, IRF-3, IRF-5, IRF-7, mitochondrial antiviral signaling molecule [MAVS], and IFN receptor [IFNAR]). We also assessed the effects of expression of these factors by mouse primary hepatocytes on HCV replication. RESULTS: In addition to IRF-3- and IFN-mediated antiviral responses, IFN-independent, but IRF-1- and IRF-5-dependent mechanisms, restrict HCV replication in mouse embryonic fibroblasts. In primary hepatocytes these IFN-independent require MAVS and IRF-1. CONCLUSIONS: HCV replication is limited by interferon-mediated pathways as well pathways that are independent of type I IFNs. IRF1 and IRF5 control IFN-independent signaling events that lead to antiviral responses. We observed antiviral roles of IRF1 and IRF5 that were IFN-independent and cell-type specific. These mechanisms are important in controlling viruses that interfere with the IFN signaling because cells retain the ability to induce functional but local antiviral states through expression of interferon-stimulated genes.


Subject(s)
Fibroblasts/virology , Hepacivirus/physiology , Hepatocytes/virology , Interferons/physiology , Signal Transduction/physiology , Virus Replication/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Antiviral Agents/therapeutic use , Fibroblasts/pathology , Hepatitis C/drug therapy , Hepatocytes/pathology , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/physiology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/physiology
14.
Cancer Res ; 71(20): 6410-8, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21900395

ABSTRACT

IFN-γ promotes tumoral immune surveillance, but its involvement in controlling metastases is less clear. Using a mouse model of pulmonary metastases, we show that local IFN-γ treatment inhibits formation of metastases through its regulation of IRF-1 in tumor cells. IRF-1 is an IFN-γ-induced transcription factor pivotal in the regulation of infection and inflammation. IRF-1 blockade abolished the inhibitory effect of IFN-γ on tumor metastases, whereas ectopic expression of IRF-1 phenocopied the inhibitory effects of IFN-γ. IRF-1 did not affect the survival of tumor cells in the circulation or their infiltration into lungs, but it was essential to support the pulmonary attraction and activation of natural killer (NK) cells. Depleting NK cells from mice abolished the protective effect of IFN-γ or IRF-1 on metastases. In addition, cytotoxicity assays revealed that tumor cells expressing IRF-1 were targeted more effectively by NK cells than IRF-1 nonexpressing tumor cells. Moreover, NK cells isolated from lungs inoculated with IRF-1-expressing tumor cells exhibit a greater cytotoxic activity. Mechanistic investigations revealed that IRF-1-induced NK cell cytotoxicity was independent of perforin and granzyme B but dependent on the NK cell activating receptor DNAM-1. Taken together, our findings establish IRF-1 as an essential mediator of the cross-talk between tumor cells and NK cells that mediate immune surveillance in the metastatic niche.


Subject(s)
Carcinoma/immunology , Colonic Neoplasms/immunology , Interferon Regulatory Factor-1/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Carcinoma/drug therapy , Carcinoma/secondary , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Granzymes/pharmacology , Interferon Regulatory Factor-1/antagonists & inhibitors , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/immunology , Killer Cells, Natural/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Perforin/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Receptors, Virus/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...